AU2020402994A1 - Methods of treating cancer - Google Patents

Methods of treating cancer Download PDF

Info

Publication number
AU2020402994A1
AU2020402994A1 AU2020402994A AU2020402994A AU2020402994A1 AU 2020402994 A1 AU2020402994 A1 AU 2020402994A1 AU 2020402994 A AU2020402994 A AU 2020402994A AU 2020402994 A AU2020402994 A AU 2020402994A AU 2020402994 A1 AU2020402994 A1 AU 2020402994A1
Authority
AU
Australia
Prior art keywords
cancer
gpa
pharmaceutically acceptable
acceptable salt
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
AU2020402994A
Inventor
David M. Darst Jr.
Foster Casimir Gonsalves
Isabel KURTH
Masoud Fakhr Tavazoie
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Inspirna Inc
Original Assignee
Inspirna Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Inspirna Inc filed Critical Inspirna Inc
Publication of AU2020402994A1 publication Critical patent/AU2020402994A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Abstract

The invention features methods of treating cancer with β-GPA. The disclosure also provides methods of treating cancer including combinations of β-GPA and additional anti-cancer therapies.

Description

METHODS OF TREATING CANCER
Background β-Guanidinopropionic acid (β-GPA), also referred to as guanidinopropionic acid, beta- guanidinopropionic acid or, N-(aminoiminomethyl)-beta-alanine is a creatine analog. Studies on animals (rats, monkeys, hamsters) show that acidic guanidine derivatives such as β-GPA can ameliorate hyperglycemia in animal models of noninsulin-dependent diabetes. Accordingly, it is sometimes used as a dietary supplement in diabetic patients to regulate blood sugar levels. β-GPA has recently been found to be effective for the suppression of metastasis, particularly liver metastasis in gastrointestinal cancers, e.g., see International Patent Publication WO2014/071067. Accordingly, the development of dosing regimens of β-GPA which result in efficacy while reducing adverse events for the treatment of cancer are needed.
Summary of the Invention
The invention features methods of treating cancer by administering about 1 ,500 mg to about 4,000 mg of β-GPA twice daily. The inventors have discovered that this dosing regimen surprisingly results in higher than expected levels of systemically circulating β-GPA.
Accordingly, in one aspect, the invention features a method of treating cancer (e.g., gastrointestinal cancer such as colon cancer or gastric cancer, pancreatic cancer, liver cancer, breast cancer, prostate cancer, lung cancer, adenocarcinoma of the esophagogastric junction, and melanoma) in a subject in need thereof. This method includes administering about 1 ,500 mg to about 4,000 mg (e.g., about 1 ,500 mg to about 2,000 mg, about 1 ,750 mg to about 2,250 mg, about 2,000 mg to about 2,500 mg, about 2,250 mg to about 2,750 mg, about 2,400 mg to about 2,800 mg, about 2,700 mg to about 3,000 mg, about 2,750 mg to about 3,250 mg, about 3,100 mg to about 3,400 mg, about 3,200 mg to about 3,600 mg) of β-GPA, or a pharmaceutically acceptable salt thereof to the subject twice per day. In some embodiments of any of the foregoing methods, the method includes administering between about 2,400 mg and about 3,600 mg of β-GPA, or a pharmaceutically acceptable salt thereof to the subject twice per day. In some embodiments of any of the foregoing methods, the method includes administering about 2,400 mg of β-GPA, or a pharmaceutically acceptable salt thereof to the subject twice per day. In some embodiments of any of the foregoing methods, the method includes administering about 3,600 mg of β-GPA, or a pharmaceutically acceptable salt thereof to the subject twice per day.
In some embodiments of any of the foregoing methods, the method further includes administering one or more further anti-cancer therapies (e.g., radiation therapy, surgery, and/or one or more therapeutic agents). In some embodiments of any of the foregoing methods, the one or more further anti-cancer therapies includes folinic acid, fluorouracil, irinotecan, and/or oxaliplatin. In some embodiments of any of the foregoing methods, the one or more further anti-cancer therapies includes FOLFIRI, i.e. , folinic acid, fluorouracil, and irinotecan. In some embodiments of any of the foregoing methods, the method includes administering about 180 mg/m2 of irinotecan intravenously over 90 minutes concurrently with about 400 mg/m2 or 2 x 250 mg/m2 folinic acid intravenously over 120 minutes followed by an optional about 400-500 mg/m2 (e.g., about 400 mg/m2) bolus of fluorouracil intravenously followed by an about 2400- 3000 mg/m2 (e.g., about 2400 mg/m2) infusion of fluorouracil intravenously over 46 hours on days 1 and 15 of each 28-day cycle, e.g., repeated about every fourteen days. In some embodiments of any of the foregoing methods, the one or more therapeutic agents is cyclocreatine, a RNAi agent, a nucleic acid, a vector, 5-fluorouracil, oxaliplatin, irinotecan, capecitabine, gemcitabine, cetuximab, taxol, avast! n, folinic acid (leucovorin), regorafenib, zaltrap, topoisomerase I inhibitors, etirinotecan pegol, tivantinib, sonolisib, sorafenib, linifanib, kinase inhibitors, telatinib, BMS-908662 (i.e., methyl N-[6-[2-(5-chloro-2- methylphenyl)-1-hydroxy-3-oxoisoindol-1-yl]-1H-benzimidazol-2-yl]carbamate), robatumumab, and/or IGF1-R inhibitors.
In some embodiments of any of the foregoing methods, the method further includes surgery (e.g., prior to, or subsequent to, administration of β-GPA, or a pharmaceutically acceptable salt thereof).
In some embodiments of any of the foregoing methods, the cancer is metastatic cancer (e.g., metastatic gastrointestinal cancer such as metastatic colon cancer or metastatic gastric cancer, metastatic pancreatic cancer, metastatic liver cancer, metastatic breast cancer, metastatic prostate cancer, metastatic lung cancer, metastatic adenocarcinoma of the esophagogastric junction, or metastatic melanoma).
In some embodiments of any of the foregoing methods, the cancer is gastrointestinal cancer (e.g., colorectal cancer, gastric cancer, or adenocarcinoma of the esophagogastric junction).
In some embodiments of any of the foregoing methods, the cancer expresses CKB. In some embodiments of any of the foregoing methods, the subject is identified to have, or to be at risk of having, metastatic cancer (e.g., on the basis of the expression level of CKB being above a predetermined reference value).
In some embodiments of any of the foregoing methods, the cancer is resistant to one or more therapeutic agents. In some embodiments of any of the foregoing methods, the cancer progressed on or after treatment with one or more anti-cancer therapies.
In some embodiments of any of the foregoing methods, the β-GPA, or a pharmaceutically acceptable salt thereof is the succinate salt of β-GPA (e.g., the 2:1 succinate salt of β-GPA).
Definitions
As used herein, the term “about” represents a value that is in the range of ±10% of the value that follows the term “about.”
As used herein, the term “administration” refers to the administration of a composition (e.g., a compound or a preparation that includes a compound as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) may be by any appropriate route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, and vitreal.
The term “cancer” refers to any cancer caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, and lymphomas. A cancer “determined to be drug resistant,” as used herein, refers to a cancer that is drug resistant, based on unresponsiveness or decreased responsiveness to a chemotherapeutic agent, or is predicted to be drug resistant based on a prognostic assay (e.g., a gene expression assay).
By a “drug resistant” cancer is meant a cancer that does not respond, or exhibits a decreased response to, one or more chemotherapeutic agents (e.g., any agent described herein).
As used herein, the term “failed to respond to a prior therapy” or “refractory to a prior therapy,” refers to a cancer that progressed despite treatment with the therapy.
As used herein, “metastatic tumor” refers to a tumor or cancer in which the cancer cells forming the tumor have a high potential to or have begun to, metastasize, or spread from one location to another location or locations within a subject, via the lymphatic system or via hematogenous spread, for example, creating secondary tumors within the subject. Such metastatic behavior may be indicative of malignant tumors. In some cases, metastatic behavior may be associated with an increase in cell migration and/or invasion behavior of the tumor cells.
Examples of cancers that can be defined as metastatic include but are not limited to non-small cell lung cancer (e.g., non-squamous non-small cell lung cancer), breast cancer, ovarian cancer, colorectal cancer, biliary tract cancer, bladder cancer, brain cancer including glioblastomas and medulloblastomas, cervical cancer, choriocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, hematological neoplasms, multiple myeloma, leukemia, intraepithelial neoplasms, liver cancer, lymphomas, neuroblastomas, oral cancer, pancreatic cancer, prostate cancer, sarcoma, skin cancer including melanoma, basocellular cancer, squamous cell cancer, testicular cancer, stromal tumors, germ cell tumors, thyroid cancer, and renal cancer.
As used herein, the term “pharmaceutical composition” refers to an active compound, formulated together with one or more pharmaceutically acceptable carriers. In some embodiments, active compound is present in unit dose amount appropriate for administration in a therapeutic regimen that shows a statistically significant probability of achieving a predetermined therapeutic effect when administered to a relevant population. In some embodiments, pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin, lungs, or oral cavity; intravaginally or intrarectally, for example, as a pessary, cream, or foam; sublingually; ocularly; transdermally; or nasally, pulmonary, and to other mucosal surfaces.
A “pharmaceutically acceptable excipient,” as used herein, refers any inactive ingredient (for example, a vehicle capable of suspending or dissolving the active compound) having the properties of being nontoxic and non-inflammatory in a subject. Typical excipients include, for example: antiadherents, antioxidants, binders, coatings, compression aids, d is integrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, or waters of hydration. Those of ordinary skill in the art are familiar with a variety of agents and materials useful as excipients.
The term “pharmaceutically acceptable salt,” as use herein, refers to those salts of the compounds described here that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley- VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.
The compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention be prepared from inorganic or organic bases. Frequently, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases are well-known in the art.
The term “subject,” as used herein, refers to a human or non-human animal (e.g., a mammal such as a non-human primate, horse, cow, or dog).
The term “treatment” (also “treat” or “treating”), in its broadest sense, refers to any administration of a substance (e.g., provided compositions) that partially or completely alleviates, ameliorates, relieves, inhibits, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms, features, and/or causes of a particular disease, disorder, and/or condition. In some embodiments, such treatment may be administered to a subject who does not exhibit signs of the relevant disease, disorder and/or condition and/or of a subject who exhibits only early signs of the disease, disorder, and/or condition. Alternatively, or additionally, in some embodiments, treatment may be administered to a subject who exhibits one or more established signs of the relevant disease, disorder and/or condition. In some embodiments, treatment may be of a subject who has been diagnosed as suffering from the relevant disease, disorder, and/or condition. In some embodiments, treatment may be of a subject known to have one or more susceptibility factors that are statistically correlated with increased risk of development of the relevant disease, disorder, and/or condition.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present disclosure; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. Detailed Description of the Invention
The invention features methods of treating cancer by administering about 2,000 mg to about 4,000 mg of β-GPA twice daily. The inventors have discovered that this dosing regimen surprisingly results in higher than expected levels of circulating β-GPA. β-GPA β-GPA has the structure: β-GPA is zwitterionic and highly soluble in water (> 50 mg/ml_), but has low solubility in organic solvents. β-GPA possesses a basic guanidino group, and is thus capable of forming both 1 :1 (β- GPA:acid) and 2:1 (β-GPA:acid) salts with diacids. As used herein, a “2:1 salt” of β-GPA with a diacid, e.g., a 2:1 succinate salt, refers to a salt including two molecules of β-GPA and one molecule of the diacid, e.g., a “2:1 succinate salt” includes two molecules of β-GPA and one molecule of succinic acid.
Treatment Methods β-GPA has recently been found to be effective for the suppression of metastasis. The mechanism of action has been hypothesized as inhibition of creatine transport and/or creatine kinase.
The phosphocreatine system promotes metastasis by enhancing the survival of disseminated cancer cells in the liver by acting as an energetic store for ATP generation to endure hepatic hypoxia. Inhibition of creatine transport into cancer cells limits the amount of phosphocreatine available to use in the production of ATP. Inhibition of creatine kinase inhibits the production of ATP through conversion of phosphocreatine to creatine.
Typical vascularized tumors that can be treated with the methods of the invention include solid tumors, particularly carcinomas, which require a vascular component for the provision of oxygen and nutrients. Exemplary solid tumors include, but are not limited to, carcinomas of the lung, breast, bone, ovary, stomach, pancreas, larynx, esophagus, testes, liver, parotid, biliary tract, colon, rectum, cervix, uterus, endometrium, kidney, bladder, prostate, thyroid, squamous cell carcinomas, adenocarcinomas, small cell carcinomas, melanomas, gliomas, glioblastomas, neuroblastomas, Kaposi's sarcoma, and sarcomas.
Treating cancer can result in a reduction in size or volume of a tumor. For example, after treatment, tumor size is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) relative to its size prior to treatment. Size of a tumor may be measured by any reproducible means of measurement. For example, the size of a tumor may be measured as a diameter of the tumor.
Treating cancer may further result in a decrease in number of tumors. For example, after treatment, tumor number is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) relative to number prior to treatment. Number of tumors may be measured by any reproducible means of measurement, e.g., the number of tumors may be measured by counting tumors visible to the naked eye or at a specified magnification (e.g., 2x, 3x, 4x, 5x, 10x, or 50x). Treating cancer can result in a decrease in number of metastatic nodules in other tissues or organs distant from the primary tumor site. For example, after treatment, the number of metastatic nodules is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) relative to number prior to treatment. The number of metastatic nodules may be measured by any reproducible means of measurement. For example, the number of metastatic nodules may be measured by counting metastatic nodules visible to the naked eye or at a specified magnification (e.g., 2x, 10x, or 50x).
Treating cancer can result in an increase in average survival time of a population of subjects treated according to the present invention in comparison to a population of untreated subjects. For example, the average survival time is increased by more than 30 days (more than 60 days, 90 days, or 120 days). An increase in average survival time of a population may be measured by any reproducible means. An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with the compound of the invention. An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.
Treating cancer can also result in a decrease in the mortality rate of a population of treated subjects in comparison to an untreated population. For example, the mortality rate is decreased by more than 2% (e.g., more than 5%, 10%, or 25%). A decrease in the mortality rate of a population of treated subjects may be measured by any reproducible means, for example, by calculating for a population the average number of disease-related deaths per unit time following initiation of treatment with a pharmaceutically acceptable salt of the invention. A decrease in the mortality rate of a population may also be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following completion of a first round of treatment with a method of the invention.
Compositions
Within the scope of this invention is a composition that contains a suitable excipient and one or more of the pharmaceutically acceptable salts described above. The composition can be a pharmaceutical composition that contains a pharmaceutically acceptable excipient, a dietary composition that contains a dietarily acceptable suitable excipient, or a cosmetic composition that contains a cosmetically acceptable excipient.
The term “pharmaceutical composition” refers to the combination of an active agent with a excipient, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo. A “pharmaceutically acceptable excipient,” after administered to or upon a subject, does not cause undesirable physiological effects. The excipient in the pharmaceutical composition must be “acceptable” also in the sense that it is compatible with the active ingredient and can be capable of stabilizing it. One or more solubilizing agents can be utilized as pharmaceutical excipients for delivery of an active compound. Examples of a pharmaceutically acceptable excipient include, but are not limited to, biocompatible vehicles, adjuvants, additives, and diluents to achieve a composition usable as a dosage form. As described above, the pharmaceutical compositions of the present invention additionally include a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, and lubricants, as suited to the particular dosage form desired. Remington’s Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional excipient medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention.
The above-described composition, in any of the forms described above, can be used for treating cancer, or any other disease or condition described herein. An effective amount refers to the amount of an active compound/agent that is required to confer a therapeutic effect on a treated subject. Effective doses will vary, as recognized by those skilled in the art, depending on the types of diseases treated, route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatment.
A pharmaceutical composition of this invention can be administered parenterally, orally, nasally, rectally, topically, or buccally. The term “parenteral” as used herein refers to subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional, or intracranial injection, as well as any suitable infusion technique.
A sterile injectable composition can be a solution or suspension in a non-toxic parenterally acceptable diluent or solvent. In addition, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides). Other commonly used surfactants, such as, but not limited to, Tweens or Spans or other similar emulsifying agents or bioavailability enhancers, which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms also can be used for the purpose of formulation.
A composition for oral administration can be any orally acceptable dosage form including capsules, tablets, emulsions and aqueous suspensions, dispersions, and solutions. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added.
Combination Therapies
In some embodiments, the pharmaceutical composition may further include an additional compound having antiproliferative activity. It will also be appreciated that the compounds and pharmaceutical compositions of the present invention can be formulated and employed in combination therapies, that is, the compounds and pharmaceutical compositions can be formulated with or administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. The particular combination of therapies (therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics and/or procedures and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder, or they may achieve different effects (e.g., control of any adverse effects).
EXAMPLES
Example 1. β-GPA Pharmacokinetics.
Method: Subjects were administered RGX-202 (a highly compressible salt form of β-GPA) in a regimen of 600 mg BID, 1 ,200 mg BID, 2,400 mg BID, or 3,600 mg BID, and plasma samples were taken from the subjects and tested using the protocol described below for levels of β-GPA over the course of 24 hours after administration.
The analyte, β-GPA, and internal standard (IS), [13C415N3]- β-GPA, are extracted from 50.0 μL of human plasma by a protein precipitation extraction procedure. The extraction procedure begins with the addition of 50.0 μL of internal standard working solution to all wells except double blanks, which receive 50.0 μL of water. Next the plate is covered and vortexed. Then 500 μL of acetonitrile/methanol (50/50, v/v) is added to all wells. Next the plate is covered, vortexed and centrifuged. Using a Tomtec Quadra 4, 200 μL of the supernatant is transferred into a new plate. Then 300 μL acetonitrile/methanol (50/50, v/v) is added to all wells. The plate is then sealed and vortexed. The extracts are chromatographed under reverse phase conditions on a Luna HPLC column 50 x 2.0 mm, 3 pm column using a gradient system with 10mM ammonium acetate in water and acetonitrile. The compounds are detected and quantified by tandem mass spectrometry in positive ion mode on a MDS Sciex API 4000 equipped with a Turbo lonspray® interface.
Results: As shown (by dose normalized accumulation ratios) in Table 1 below, administration with 2,400 mg BID or 3,600 mg BID of β-GPA results in higher than expected AUC and Cmax levels in the subjects.
Table 1.
Example 2. β-GPA for the Treatment of Gl Tumors.
Method: Subjects with advanced gastrointestinal tumors (e.g., locally advanced and unresectable, or metastatic) were administered multiple doses of orally administered RGX-202 as a monotherapy or in combination with irinotecan, folinic acid, and fluorouracil. In the monotherapy arm of the study, RGX-202 was administered orally twice or three times daily on days 1-28 of each 28-day cycle. The dose regimen was dependent on the cohort in which the patient was enrolled. In the combination arm of the study, RGX-202 was administered in the same way as described for the monotherapy arm in combination with FOLFIRI. FOLFIRI was administered by intravenous administration of irinotecan (180 mg/m2) over 90 minutes concurrently with intravenous administration of folinic acid (400 mg/m2) over 2 hours, followed by fluorouracil (5-FU) (400mg/m2) intravenous bolus and then 5-FU (2400mg/m2) intravenous infusion over 46 hours, on days 1 and 15 of each 28-day cycle.
During the dose escalation, subjects were evaluated for pharmacokinetics (using the method as described in Example 1), pharmacodynamics, safety, and efficacy of the drug.
Results: The subjects did not exhibit dose limiting toxicity. Objective monotherapy (RGX-202) and combination therapy (RGX-202 +FOLFIRI) activities were observed. Of the 7 subjects who received the combination therapy, 6 subjects showed stable disease (as described by RECIST 1.1 guidelines) after 40 weeks of treatment (e.g., doses of RGX-202 > 1 ,800 mg BID). Of the 10 subjects who received the monotherapy, one subject exhibited a partial response (at a dose of 3,600 mg BID after 40 weeks), and three exhibited stable disease (at doses of 1 ,200 mg BID, 2,400 mg BID, and 3,600 mg BID).
Other Embodiments
While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features herein before set forth. This application claims the benefit of U.S. provisional serial no. 62/946,581 , filed December 11 , 2019, which is incorporated herein in its entirety.

Claims (21)

What is claimed is: CLAIMS
1. A method of treating cancer in a subject in need thereof, the method comprising administering about 1 ,500 mg to about 4,000 mg of β-guanidinopropionic acid (β-GPA), or a pharmaceutically acceptable salt thereof to the subject twice per day.
2. The method of claim 1 , wherein the method comprises administering between about 2,400 mg and about 3,600 mg of β-GPA, or a pharmaceutically acceptable salt thereof to the subject twice per day.
3. The method of claim 1 or 2, wherein the method comprises administering about 2,400 mg of β-GPA, or a pharmaceutically acceptable salt thereof to the subject twice per day.
4. The method of claim 1 or 2, wherein the method comprises administering about 3,600 mg of β-GPA, or a pharmaceutically acceptable salt thereof to the subject twice per day.
5. The method of any one of claims 1 to 4, wherein the method further comprises administering one or more further anti-cancer therapies.
6. The method of claim 5, wherein the one or more anti-cancer therapies comprise radiation therapy, surgery, and/or one or more therapeutic agents.
7. The method of claim 5 or 6, wherein the one or more further anti-cancer therapies comprise folinic acid, fluorouracil, irinotecan, and/or oxaliplatin.
8. The method of any one of claims 5 to 7, wherein the one or more further anti-cancer therapies comprise folinic acid, fluorouracil, and irinotecan.
9. The method of claim 8, wherein method comprises administering about 180 mg/m2 of irinotecan intravenously over 90 minutes concurrently with about 400 mg/m2 folinic acid intravenously over 120 minutes followed by an about 2400 mg/m2 infusion of fluorouracil intravenously over 46 hours.
10. The method of claim 9, wherein the administering is repeated about every fourteen days,
11. The method of any one of claims 5 to 10, wherein the method further comprises surgery.
12. The method of claim 11 , wherein the surgery is prior to administration of β-GPA, or a pharmaceutically acceptable salt thereof.
13. The method of any one of claims 1 to 12, wherein the cancer is metastatic cancer.
14. The method of any one of claims 1 to 13, wherein the cancer is gastrointestinal cancer.
15. The method of claim 14, wherein the gastrointestinal cancer is colorectal cancer, gastric cancer, or adenocarcinoma of the esophagogastric junction.
16. The method of any one of claims 1 to 15, wherein the cancer expresses CKB.
17. The method of any one of claims 1 to 16, wherein subject is identified to have, or to be at risk of having, metastatic cancer.
18. The method of any one of claims 1 to 17, wherein the cancer is resistant to one or more therapeutic agents.
19. The method of any one of claims 1 to 18, wherein the cancer progressed on or after treatment with one or more anti-cancer therapies.
20. The method of any one of claims 1 to 19, wherein the β-GPA, or a pharmaceutically acceptable salt thereof is the succinate salt of β-GPA.
21. The method of claim 20, wherein the succinate salt of β-GPA is the 2:1 succinate salt of β-
GPA.
AU2020402994A 2019-12-11 2020-12-10 Methods of treating cancer Pending AU2020402994A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201962946581P 2019-12-11 2019-12-11
US62/946,581 2019-12-11
PCT/US2020/064317 WO2021119316A1 (en) 2019-12-11 2020-12-10 Methods of treating cancer

Publications (1)

Publication Number Publication Date
AU2020402994A1 true AU2020402994A1 (en) 2022-06-23

Family

ID=76316395

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2020402994A Pending AU2020402994A1 (en) 2019-12-11 2020-12-10 Methods of treating cancer

Country Status (8)

Country Link
US (1) US20210177790A1 (en)
EP (1) EP4072561A4 (en)
JP (1) JP2023505687A (en)
AU (1) AU2020402994A1 (en)
CA (1) CA3161341A1 (en)
IL (1) IL293721A (en)
TW (1) TW202135792A (en)
WO (1) WO2021119316A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102615692B1 (en) * 2021-10-13 2023-12-19 주식회사 휴엔켐 Composition for preventing or treating liver fibrosis comprising beta-guanidinopropionic acid

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK0601001T3 (en) * 1991-08-26 1997-10-20 Upjohn Co Liquid food product containing 3-guanidinopropionic acid.
AU2013337768B2 (en) * 2012-10-31 2015-07-02 The Rockefeller University Treatment and diagnosis of colon cancer
CN107708700A (en) * 2015-04-17 2018-02-16 波士顿生物医药有限公司 Method for treating cancer
US20170056352A1 (en) * 2015-08-25 2017-03-02 Rgenix, Inc. PHARMACEUTICALLY ACCEPTABLE SALTS OF beta-GUANIDINOPROPIONIC ACID WITH IMPROVED PROPERTIES AND USES THEREOF
WO2018160178A1 (en) * 2017-03-01 2018-09-07 Rgenix, Inc. Pharmaceutically acceptable salts of b-guanidinopropionic acid with improved properties and uses thereof

Also Published As

Publication number Publication date
US20210177790A1 (en) 2021-06-17
EP4072561A4 (en) 2023-11-29
IL293721A (en) 2022-08-01
EP4072561A1 (en) 2022-10-19
JP2023505687A (en) 2023-02-10
CA3161341A1 (en) 2021-06-17
WO2021119316A1 (en) 2021-06-17
TW202135792A (en) 2021-10-01

Similar Documents

Publication Publication Date Title
AU2002210993B2 (en) Medicinal compositions for concominant use as anticancer agents
TWI399206B (en) Antitumor agent
DK2799070T3 (en) EFFECTIVE AMPLIFIER FOR ANTITUMUM AGENTS
EP3443962A1 (en) Quinoline derivative for treating gastric cancer
EP3429582B1 (en) Combination therapy for proliferative diseases
RU2752172C2 (en) Antitumor agent, antitumor enhancer and antitumor kit
WO2010086964A1 (en) Combination therapy for treating cancer
JPWO2020260252A5 (en)
WO2019176984A1 (en) Antitumor agent, antitumor effect potentiator and antitumor kit
EP4072561A1 (en) Methods of treating cancer
US11141421B2 (en) Antitumor agent for biliary tract cancer and method for treating biliary tract cancer
EP3429572B1 (en) Combination therapy for proliferative diseases
US20190231793A1 (en) Use of harringtonines in the treatment of breast cancer, in particular triple-negative breast cancer
JP6973456B2 (en) Side effect reducing agent during cancer chemotherapy
PT1604991E (en) Antitumor effect potentiator and antitumor agent
TW202114682A (en) Combination of a mcl-1 inhibitor and a standard of care treatment for breast cancer, uses and pharmaceutical compositions thereof
WO2007132221A1 (en) Combined anticancer pyrimidine-thiazole aurora kinase inhibitors
EP2268287B1 (en) Dosage regimens of an antitumor agent comprising deoxycytidine derivative
EP3492082B1 (en) Anticancer pharmaceutical composition
US20100087398A1 (en) Dihydropyridine derivative for treating cancer or a pre-cancerous condition and other conditions
JP5409613B2 (en) Antitumor agent, kit and cancer treatment method
TW201728328A (en) Method for treating cancer patients with severe renal impairment
EP3804721A1 (en) Antitumor agent and method for tumor therapy
JP2021011453A (en) Agent for treating or preventing skin disorder caused by anticancer agent administration
CN117298148A (en) Application of aminodithioformate compound or pharmaceutically acceptable salt thereof in combination with copper ions in preparation of antitumor drugs